Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Chem Biol Drug Des ; 103(1): e14361, 2024 01.
Article in English | MEDLINE | ID: mdl-37767622

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is a homo-trimeric protein complex that clamps around DNA to tether DNA polymerases to the template during replication and serves as a hub for many other interacting proteins. It regulates DNA metabolic processes and other vital cellar functions through the binding of proteins having short linear motifs (SLiMs) like the PIP-box (PCNA-interacting protein-box) or the APIM (AlkB homolog 2 PCNA-interacting motif) in the hydrophobic pocket where SLiMs bind. However, overproducing TbPCNA or human PCNA (hPCNA) in the pathogenic protist Trypanosoma brucei triggers a dominant-negative phenotype of arrested proliferation. The mechanism for arresting T. brucei proliferation requires the overproduced PCNA orthologs to have functional intact SLiM-binding pocket. Sight-directed mutagenesis studies showed that T. brucei overproducing PCNA variants with disrupted SLiM-binding pockets grew normally. We hypothesized that chemically disrupting the SLiM-binding pocket would restore proliferation in T. brucei, overproducing PCNA orthologs. Testing this hypothesis is the proof-of-concept for a T. brucei-based PCNA screening assay. The assay design is to discover bioactive small molecules that restore proliferation in T. brucei strains that overproduce PCNA orthologs, likely by disrupting interactions in the SLiM-binding pocket. The pilot screen for this assay discovered two hit compounds that linked to predetermined PCNA targets. Compound #1, a known hPCNA inhibitor, had selective bioactivity to hPCNA overproduced in T. brucei, validating the assay. Compound #6 had promiscuous bioactivity for hPCNA and TbPCNA but is the first compound discovered with bioactivity for inhibiting TbPCNA.


Subject(s)
DNA Replication , Trypanosoma brucei brucei , Humans , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Trypanosoma brucei brucei/metabolism , DNA/metabolism , Mutagenesis , Protein Binding
2.
Proc Natl Acad Sci U S A ; 116(48): 24214-24220, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31723045

ABSTRACT

Although CRIg was originally identified as a macrophage receptor for binding complement C3b/iC3b in vitro, recent studies reveal that CRIg functions as a pattern recognition receptor in vivo for Kupffer cells (KCs) to directly bind bacterial pathogens in a complement-independent manner. This raises the critical question of whether CRIg captures circulating pathogens through interactions with complement in vivo under flow conditions. Furthermore, the role of CRIg during parasitic infection is unknown. Taking advantage of intravital microscopy and using African trypanosomes as a model, we studied the role of CRIg in intravascular clearance of bloodborne parasites. Complement C3 is required for intravascular clearance of African trypanosomes by KCs, preventing the early mortality of infected mice. Moreover, antibodies are essential for complement-mediated capture of circulating parasites by KCs. Interestingly, reduced antibody production was observed in the absence of complement C3 during infection. We further demonstrate that CRIg but not CR3 is critically involved in KC-mediated capture of circulating parasites, accounting for parasitemia control and host survival. Of note, CRIg cannot directly catch circulating parasites and antibody-induced complement activation is indispensable for CRIg-mediated parasite capture. Thus, we provide evidence that CRIg, by interacting with complement in vivo, plays an essential role in intravascular clearance of bloodborne parasites. Targeting CRIg may be considered as a therapeutic strategy.


Subject(s)
Complement C3b/metabolism , Host-Parasite Interactions/physiology , Parasitemia/parasitology , Receptors, Complement/physiology , Trypanosomiasis, African/blood , Animals , Complement C3b/immunology , Intravital Microscopy , Kupffer Cells/immunology , Kupffer Cells/parasitology , Macrophage-1 Antigen/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/pathogenicity , Trypanosoma congolense/pathogenicity , Trypanosomiasis, African/mortality , Trypanosomiasis, African/parasitology
3.
Mol Biochem Parasitol ; 220: 1-4, 2018 03.
Article in English | MEDLINE | ID: mdl-29287675

ABSTRACT

Trypanosoma brucei sub-species are vector borne kinetoplastid parasites that cause the potentially lethal disease Human African trypanosomiasis. The target-based therapy for curing this parasitic disease relies on one drug, Eflornithine. The roles of mitogen-activated protein kinases in regulating key cellular processes in eukaryotic cells such as proliferation, stress response and differentiation plus their druggability make them attractive targets for therapeutic exploitation. The extracellular-regulated kinase 8 homolog in T. brucei (TbERK8) is a MAPK that is required for the parasite to proliferate normally in culture. We examined the importance of TbERK8 for permitting T. brucei to thrive in mice. Here we show that depleting TbERK8 in vivo negatively affected the virulence of T. brucei reducing its ability to progress to lethal infections or cause significant pathology in mice, which validates it as an attractive target.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/genetics , Gene Deletion , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/parasitology , Virulence Factors/genetics , Animals , Disease Models, Animal , Mice , Survival Analysis , Trypanosoma brucei brucei/genetics , Virulence
4.
Cell Cycle ; 15(20): 2827-41, 2016 Oct 17.
Article in English | MEDLINE | ID: mdl-27589575

ABSTRACT

The Trypanosoma brucei subspecies T. brucei gambiense and T. brucei rhodesiense are vector-borne pathogens that cause sleeping sickness also known as Human African Trypanosomiasis (HAT), which is fatal if left untreated. The drugs that treat HAT are ineffective and cause toxic side effects. One strategy for identifying safer and more effective HAT drugs is to therapeutically exploit essential gene targets in T. brucei. Genes that make up a basic mitogen-activated protein kinase (MAPK) network are present in T. brucei. Tb927.10.5140 encodes an essential MAPK that is homologous to the human extracellular-signal regulated kinase 8 (HsERK8) which forms a tight complex with the replication factor proliferating cell nuclear antigen (PCNA) to stabilize intracellular PCNA levels. Here we demonstrate that (TbPCNA) is uniquely phos-phorylated on serine (S) and threonine (T) residues in T. brucei and that TbERK8 phosphorylates TbPCNA at each of these residues. The ability of an ERK8 homolog to phosphorylate a PCNA homolog is a novel biochemical property that is first demonstrated here in T. brucei and may be unique to this pathogen. We demonstrate that the potent HsERK8 inhibitor Ro318220, has an IC50 for TbERK8 that is several hundred times higher than its reported IC50 for HsERK8. This indicated that the active sites of TbERK8 and HsERK8 can be selectively inhibited, which provides a rational basis for discovering inhibitors that specifically target this essential parasite MAPK to kill the parasite.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Sequence Homology, Amino Acid , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Catalytic Domain , Extracellular Signal-Regulated MAP Kinases/chemistry , Humans , Immunoprecipitation , Indoles/pharmacology , Phosphorylation/drug effects , Phylogeny , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/metabolism , Sequence Alignment , Sf9 Cells , Small Molecule Libraries/pharmacology , Trypanosoma brucei brucei/drug effects
5.
Bioorg Med Chem ; 24(19): 4647-4651, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27519462

ABSTRACT

Human African trypanosomiasis (HAT) is a lethal, vector-borne disease caused by the parasite Trypanosoma brucei. Therapeutic strategies for this neglected tropical disease suffer from disadvantages such as toxicity, high cost, and emerging resistance. Therefore, new drugs with novel modes of action are needed. We screened cultured T. brucei against a focused kinase inhibitor library to identify promising bioactive compounds. Among the ten hits identified from the phenotypic screen, AZ960 emerged as the most promising compound with potent antiparasitic activity (IC50=120nM) and was shown to be a selective inhibitor of an essential gene product, T. brucei extracellular signal-regulated kinase 8 (TbERK8). We report that AZ960 has a Ki of 1.25µM for TbERK8 and demonstrate its utility in establishing TbERK8 as a potentially druggable target in T. brucei.


Subject(s)
Aminopyridines/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Aminopyridines/chemistry , Drug Discovery , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Recombinant Proteins/metabolism , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/metabolism
6.
Cell Cycle ; 14(4): 674-88, 2015.
Article in English | MEDLINE | ID: mdl-25701409

ABSTRACT

The DNA replication machinery is spatially and temporally coordinated in all cells to reproduce a single exact copy of the genome per division, but its regulation in the protozoan parasite Trypanosoma brucei is not well characterized. We characterized the effects of altering the levels of proliferating cell nuclear antigen, a key component of the DNA replication machinery, in bloodstream form T. brucei. This study demonstrated that tight regulation of TbPCNA levels was critical for normal proliferation and DNA replication in the parasite. Depleting TbPCNA mRNA reduced proliferation, severely diminished DNA replication, arrested the synthesis of new DNA and caused the parasites to accumulated in G2/M. Attenuating the parasite by downregulating TbPCNA caused it to become hypersensitive to hydroxyurea. Overexpressing TbPCNA in T. brucei arrested proliferation, inhibited DNA replication and prevented the parasite from exiting G2/M. These results indicate that distinct mechanisms of cell cycle arrest are associated with upregulating or downregulating TbPCNA. The findings of this study validate deregulating intra-parasite levels of TbPCNA as a potential strategy for therapeutically exploiting this target in bloodstream form T. brucei.


Subject(s)
Cell Cycle/physiology , Cell Proliferation/physiology , DNA Replication/physiology , G2 Phase Cell Cycle Checkpoints/physiology , Proliferating Cell Nuclear Antigen/metabolism , Trypanosoma brucei brucei/physiology , Cell Culture Techniques , DNA Primers/genetics , Flow Cytometry , Immunoblotting , Luciferases , Microscopy, Fluorescence , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trypanosoma brucei brucei/metabolism
7.
J Nat Prod ; 76(4): 630-41, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23445522

ABSTRACT

Almiramide C is a marine natural product with low micromolar activity against Leishmania donovani, the causative agent of leishmaniasis. We have now shown that almiramide C is also active against the related parasite Trypanosoma brucei, the causative agent of human African trypanosomiasis. A series of activity-based probes have been synthesized to explore both the molecular target of this compound series in T. brucei lysates and site localization through epifluorescence microscopy. These target identification studies indicate that the almiramides likely perturb glycosomal function through disruption of membrane assembly machinery. Glycosomes, which are organelles specific to kinetoplastid parasites, house the first seven steps of glycolysis and have been shown to be essential for parasite survival in the bloodstream stage. There are currently no reported small-molecule disruptors of glycosome function, making the almiramides unique molecular probes for this understudied parasite-specific organelle. Additionally, examination of toxicity in an in vivo zebrafish model has shown that these compounds have little effect on organism development, even at high concentrations, and has uncovered a potential side effect through localization of fluorescent derivatives to zebrafish neuromast cells. Combined, these results further our understanding of the potential value of this lead series as development candidates against T. brucei.


Subject(s)
Biological Products/pharmacology , Chagas Disease/drug therapy , Lipopeptides/pharmacology , Trypanosoma brucei brucei/drug effects , Animals , Biological Products/chemistry , Glycolysis/physiology , Humans , Leishmania donovani/drug effects , Microbodies/metabolism , Microscopy, Fluorescence , Trypanosoma brucei brucei/metabolism , Zebrafish/physiology
8.
PLoS One ; 7(8): e43913, 2012.
Article in English | MEDLINE | ID: mdl-22952808

ABSTRACT

Human African trypanosomiasis or sleeping sickness is a vector-borne parasitic disease that has a major impact on human health and welfare in sub-Saharan countries. Based mostly on data from animal models, it is currently thought that trypanosome entry into the brain occurs by initial infection of the choroid plexus and the circumventricular organs followed days to weeks later by entry into the brain parenchyma. However, Trypanosoma brucei bloodstream forms rapidly cross human brain microvascular endothelial cells in vitro and appear to be able to enter the murine brain without inflicting cerebral injury. Using a murine model and intravital brain imaging, we show that bloodstream forms of T. b. brucei and T. b. rhodesiense enter the brain parenchyma within hours, before a significant level of microvascular inflammation is detectable. Extravascular bloodstream forms were viable as indicated by motility and cell division, and remained detectable for at least 3 days post infection suggesting the potential for parasite survival in the brain parenchyma. Vascular inflammation, as reflected by leukocyte recruitment and emigration from cortical microvessels, became apparent only with increasing parasitemia at later stages of the infection, but was not associated with neurological signs. Extravascular trypanosomes were predominantly associated with postcapillary venules suggesting that early brain infection occurs by parasite passage across the neuroimmunological blood brain barrier. Thus, trypanosomes can invade the murine brain parenchyma during the early stages of the disease before meningoencephalitis is fully established. Whether individual trypanosomes can act alone or require the interaction from a quorum of parasites remains to be shown. The significance of these findings for disease development is now testable.


Subject(s)
Brain/parasitology , Trypanosoma brucei brucei/physiology , Animals , Blood/parasitology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/parasitology , Brain/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Species Specificity , Time Factors , Trypanosoma brucei brucei/metabolism
9.
Chem Biol Drug Des ; 78(3): 454-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21668652

ABSTRACT

New drugs are needed to treat human African trypanosomiasis because the currently approved treatments are toxic or limited in efficacy. One strategy for developing new drugs involves discovering novel genes whose products can be targeted for modulation by small-molecule chemotherapeutic agents. The Trypanosoma brucei genome contains many genes with the potential to become such targets. Kinases represent one group of genes that regulate many important cell functions and can be modulated by small molecules, thus representing a promising group of enzymes to screen for potential therapeutic targets. RNAi screens could help identify the most promising kinase targets, but the lack of suitable assays represents a barrier for optimizing the use of this technology in T. brucei. Here, we describe an RNAi screen of a small RNAi library targeting 30 members of the T. brucei kinome utilizing a luciferase-based assay. This screen both validated the luciferase-based assay as a suitable method for conducting RNAi screens in T. brucei and also identified two kinases (CRK12 and ERK8) that are essential for normal proliferation by the parasite.


Subject(s)
Drug Discovery , Phosphotransferases/genetics , RNA Interference , Trypanosoma brucei brucei/enzymology , Animals , Humans , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/drug therapy
10.
PLoS Negl Trop Dis ; 5(5): e1023, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21572521

ABSTRACT

The targeting of parasite cysteine proteases with small molecules is emerging as a possible approach to treat tropical parasitic diseases such as sleeping sickness, Chagas' disease, and malaria. The homology of parasite cysteine proteases to the human cathepsins suggests that inhibitors originally developed for the latter may be a source of promising lead compounds for the former. We describe here the screening of a unique ∼ 2,100-member cathepsin inhibitor library against five parasite cysteine proteases thought to be relevant in tropical parasitic diseases. Compounds active against parasite enzymes were subsequently screened against cultured Plasmodium falciparum, Trypanosoma brucei brucei and/or Trypanosoma cruzi parasites and evaluated for cytotoxicity to mammalian cells. The end products of this effort include the identification of sub-micromolar cell-active leads as well as the elucidation of structure-activity trends that can guide further optimization efforts.


Subject(s)
Antiparasitic Agents/isolation & purification , Antiparasitic Agents/metabolism , Cysteine Proteases/metabolism , Drug Evaluation, Preclinical , Protease Inhibitors/isolation & purification , Protease Inhibitors/metabolism , Parasitic Sensitivity Tests , Plasmodium falciparum/drug effects , Trypanosoma brucei brucei/drug effects , Trypanosoma cruzi/drug effects
11.
PLoS Negl Trop Dis ; 4(6): e701, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20544024

ABSTRACT

BACKGROUND: Trypanosoma brucei is the etiological agent of Human African Trypanosomiasis, an endemic parasitic disease of sub-Saharan Africa. TbCatB and rhodesain are the sole Clan CA papain-like cysteine proteases produced by the parasite during infection of the mammalian host and are implicated in the progression of disease. Of considerable interest is the exploration of these two enzymes as targets for cysteine protease inhibitors that are effective against T. brucei. METHODS AND FINDINGS: We have determined, by X-ray crystallography, the first reported structure of TbCatB in complex with the cathepsin B selective inhibitor CA074. In addition we report the structure of rhodesain in complex with the vinyl-sulfone K11002. CONCLUSIONS: The mature domain of our TbCat*CA074 structure contains unique features for a cathepsin B-like enzyme including an elongated N-terminus extending 16 residues past the predicted maturation cleavage site. N-terminal Edman sequencing reveals an even longer extension than is observed amongst the ordered portions of the crystal structure. The TbCat*CA074 structure confirms that the occluding loop, which is an essential part of the substrate-binding site, creates a larger prime side pocket in the active site cleft than is found in mammalian cathepsin B-small molecule structures. Our data further highlight enhanced flexibility in the occluding loop main chain and structural deviations from mammalian cathepsin B enzymes that may affect activity and inhibitor design. Comparisons with the rhodesain*K11002 structure highlight key differences that may impact the design of cysteine protease inhibitors as anti-trypanosomal drugs.


Subject(s)
Cysteine Endopeptidases/chemistry , Trypanosoma brucei brucei/chemistry , Binding Sites , Crystallography, X-Ray/methods , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Dipeptides/chemistry , Dipeptides/metabolism , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sulfones/chemistry , Sulfones/metabolism , Trypanosoma brucei brucei/enzymology
12.
Nat Prod Commun ; 4(2): 193-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19370921

ABSTRACT

The alkaloid cryptolepine (1) and eight synthetic analogues (2-8) were assessed for in vitro activities against Trypanosoma brucei. Four of the analogues were found to be highly potent with IC50 values of less than 3 nM and three of these were assessed against T. brucei brucei infection in rats. The most effective compound was 2, 7-dibromocryptolepine (7); a single oral dose of 20 mg/kg suppressed parasitaemia and increased the mean survival time to 13.6 days compared with 8.4 days for untreated controls. In addition, four huperzine derivatives (9-12) were shown to have in vitro antitrypanosomal activities with IC50 values ranging from 303-377 nM.


Subject(s)
Indole Alkaloids/chemistry , Indole Alkaloids/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Animals , Cryptolepis/chemistry , Drug Administration Routes , Humans , Huperzia/chemistry , Molecular Structure , Rats , Sesquiterpenes/administration & dosage , Trypanosomiasis, African/drug therapy
13.
Biotechnol Prog ; 25(1): 124-31, 2009.
Article in English | MEDLINE | ID: mdl-19197977

ABSTRACT

Two SDS-resistant endochitinases, designated as ASCHI53 and ASCHI61, were isolated from Aeromonas schubertii in a soil sample from southern Taiwan. MALDI-TOF mass measurement indicates the molecular weights of 53,527 for ASCHI53 and 61,202 for ASCHI61. N-terminal and internal amino acid sequences were obtained, and BLAST analysis of the sequences and MS/MS peptide sequencing showed that they were novel proteins. Degradation of chitin by these two endochitinases gave rise to hexameric chitin oligosaccharide, a compound known to have several potent biomedical functions. ASCHI53 and ASCHI61 retained, respectively, 65% and 75%, of their chitinase activity in the presence of 5% SDS and 100% of their activity in the presence of 10% beta-mercaptoethanol. These results demonstrate that they are SDS-resistant endochitinases and probably have a rigid structure.


Subject(s)
Aeromonas/enzymology , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Chitinases/isolation & purification , Chitinases/metabolism , Enzyme Activation/drug effects , Sodium Dodecyl Sulfate/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
14.
PLoS Negl Trop Dis ; 3(2): e372, 2009.
Article in English | MEDLINE | ID: mdl-19190730

ABSTRACT

BACKGROUND: The two front-line drugs for chronic Trypanosoma cruzi infections are limited by adverse side-effects and declining efficacy. One potential new target for Chagas' disease chemotherapy is sterol 14alpha-demethylase (CYP51), a cytochrome P450 enzyme involved in biosynthesis of membrane sterols. METHODOLOGY/PRINCIPAL FINDING: In a screening effort targeting Mycobacterium tuberculosis CYP51 (CYP51(Mt)), we previously identified the N-[4-pyridyl]-formamide moiety as a building block capable of delivering a variety of chemotypes into the CYP51 active site. In that work, the binding modes of several second generation compounds carrying this scaffold were determined by high-resolution co-crystal structures with CYP51(Mt). Subsequent assays against the CYP51 orthologue in T. cruzi, CYP51(Tc), demonstrated that two of the compounds tested in the earlier effort bound tightly to this enzyme. Both were tested in vitro for inhibitory effects against T. cruzi and the related protozoan parasite Trypanosoma brucei, the causative agent of African sleeping sickness. One of the compounds had potent, selective anti-T. cruzi activity in infected mouse macrophages. Cure of treated host cells was confirmed by prolonged incubation in the absence of the inhibiting compound. Discrimination between T. cruzi and T. brucei CYP51 by the inhibitor was largely based on the variability (phenylalanine versus isoleucine) of a single residue at a critical position in the active site. CONCLUSIONS/SIGNIFICANCE: CYP51(Mt)-based crystal structure analysis revealed that the functional groups of the two tightly bound compounds are likely to occupy different spaces in the CYP51 active site, suggesting the possibility of combining the beneficial features of both inhibitors in a third generation of compounds to achieve more potent and selective inhibition of CYP51(Tc).


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Mycobacterium tuberculosis/enzymology , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Trypanosoma cruzi , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Chagas Disease/drug therapy , Chagas Disease/parasitology , Cytochrome P-450 Enzyme System , Enzyme Inhibitors/adverse effects , Humans , Inhibitory Concentration 50 , Mice , Mycobacterium tuberculosis/drug effects , Parasitic Sensitivity Tests , Trypanocidal Agents/adverse effects , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology
15.
PLoS Negl Trop Dis ; 2(9): e298, 2008 Sep 24.
Article in English | MEDLINE | ID: mdl-18820745

ABSTRACT

We investigated the roles played by the cysteine proteases cathepsin B and cathepsin L (brucipain) in the pathogenesis of Trypansoma brucei brucei in both an in vivo mouse model and an in vitro model of the blood-brain barrier. Doxycycline induction of RNAi targeting cathepsin B led to parasite clearance from the bloodstream and prevent a lethal infection in the mice. In contrast, all mice infected with T. brucei containing the uninduced Trypanosoma brucei cathepsin B (TbCatB) RNA construct died by day 13. Induction of RNAi against brucipain did not cure mice from infection; however, 50% of these mice survived 60 days longer than uninduced controls. The ability of T. b. brucei to cross an in vitro model of the human blood-brain barrier was also reduced by brucipain RNAi induction. Taken together, the data suggest that while TbCatB is the more likely target for the development of new chemotherapy, a possible role for brucipain is in facilitating parasite entry into the brain.


Subject(s)
Cathepsin B/genetics , Cathepsin L/genetics , RNA Interference , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , Animals , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Genes, Reporter , Humans , Life Cycle Stages/genetics , Life Cycle Stages/physiology , Mice , Models, Animal , Plasmids , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Interference/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Tetracycline/pharmacology , Transfection , Trypanosoma brucei brucei/physiology , Trypanosomiasis, African
16.
J Biol Chem ; 283(43): 28934-43, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18701454

ABSTRACT

Cysteine proteases of the Clan CA (papain) family are the predominant protease group in primitive invertebrates. Cysteine protease inhibitors arrest infection by the protozoan parasite, Trypanosoma brucei. RNA interference studies implicated a cathepsin B-like protease, tbcatB, as a key inhibitor target. Utilizing parasites in which one of the two alleles of tbcatb has been deleted, the key role of this protease in degradation of endocytosed host proteins is delineated. TbcatB deficiency results in a decreased growth rate and dysmorphism of the flagellar pocket and the subjacent endocytic compartment. Western blot and microscopic analysis indicate that deficiency in tbcatB results in accumulation of both host and parasite proteins, including the lysosomal marker p67. A critical function for parasitism is the degradation of host transferrin, which is necessary for iron acquisition. Substrate specificity analysis of recombinant tbcatB revealed the optimal peptide cleavage sequences for the enzyme and these were confirmed experimentally using FRET-based substrates. Degradation of transferrin was validated by SDS-PAGE and the specific cleavage sites identified by N-terminal sequencing. Because even a modest deficiency in tbcatB is lethal for the parasite, tbcatB is a logical target for the development of new anti-trypanosomal chemotherapy.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine/chemistry , Iron/metabolism , Animals , Animals, Genetically Modified , Catalytic Domain , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/physiology , Endocytosis , Fluorescence Resonance Energy Transfer , Iron/chemistry , Lysosomes/metabolism , Microscopy, Electron, Transmission , Models, Biological , Protein Structure, Tertiary , RNA Interference , Transferrin/chemistry , Trypanosoma brucei brucei/metabolism
17.
Curr Pharm Des ; 14(9): 889-900, 2008.
Article in English | MEDLINE | ID: mdl-18473838

ABSTRACT

Lysosomes were first described as vacuolar structures containing various hydrolytic enzymes at acidic pH. Subsequent studies revealed that the lysosome/vacuolar system is complex and composed of distinct membrane-enclosed vesicles including endosomes, primary and mature lysosomes, autophagic vesicles, residual bodies, multivesicular bodies, and digestive lysosomes. Lysosomes express a battery of hydrolytic enzymes including proteases, acid phosphatases, glycosidases, and lipases. Parasitic protozoa also possess complex intracellular lysosomes/endosomes/vesicles involved in digestion, transport and recycling of molecules similar to those of mammalian cells. Unique characteristics are ascribed to lysosomes of different parasites and may even differ between parasite stages. Transport of hydrolases and proteins to parasite lysosomes is directed either from the Golgi complex via endosomal vesicles or from endocytic vesicles originated in the cell surface. Inhibition of lysosomal proteases demonstrated that different proteolytic machineries catabolize distinct classes of proteins, and this selectivity may be exploited for the development of effective antiparasitic drugs. This review describes lysosomal molecules that are either validated or potential drug targets for Chagas' disease, sleeping sickness, leishmaniasis, toxoplasmosis, malaria, amebiasis, and giardiasis.


Subject(s)
Eukaryota/drug effects , Lysosomes/drug effects , Protozoan Infections/drug therapy , Animals , Antiprotozoal Agents/pharmacology , Drug Delivery Systems , Eukaryota/metabolism , Humans , Lysosomes/enzymology , Lysosomes/metabolism
18.
Chem Biol Drug Des ; 67(5): 355-63, 2006 May.
Article in English | MEDLINE | ID: mdl-16784460

ABSTRACT

Chemotherapy against human African trypanosomiasis relies on four drugs that cause frequent and occasionally severe side-effects. Because human African trypanosomiasis is a disease of poor people in Africa, the traditional market-driven pathways to drug development are not available. One potentially rapid and cost-effective approach to identifying and developing new trypanocidal drugs would be high throughput-screening of existing drugs already approved for other uses, as well as clinical candidates in late development. We have developed an ATP-bioluminescence assay that could be used to rapidly and efficiently screen compound libraries against trypanosomes in a high throughput-screening format to validate this notion. We screened a collection of 2160 FDA-approved drugs, bioactive compounds and natural products to identify hits that were cytotoxic to cultured Trypanosoma brucei at a concentration of 1 mum or less. This meant that any hit identified would be effective at a concentration readily achievable by standard drug dosing in humans. From the screen, 35 hits from seven different drug categories were identified. These included the two approved trypanocidal drugs, suramin and pentamidine, several other drugs suspected but never validated as trypanocidal, and 17 novel trypanocidal drugs.


Subject(s)
Drug Evaluation, Preclinical/methods , Parasitic Sensitivity Tests/methods , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Animals , Diazomethane/analogs & derivatives , Humans , Luminescent Measurements/methods , Molecular Structure , Trypanocidal Agents/chemistry , Trypanocidal Agents/economics , Trypanosoma brucei brucei/metabolism
19.
J Med Chem ; 49(6): 2096-102, 2006 Mar 23.
Article in English | MEDLINE | ID: mdl-16539398

ABSTRACT

Treatment of the 6-N-cyclopropyl-2',3'-di-O-isopropylideneadenosine 5'-aldehyde with sulfone-stabilized phosphonate or fluorophosphonate reagents followed by stannyldesulfonylations and subsequent iodo- or protiodestannylation gave 6-N-cyclopropyl-5'-deoxy-5'-(iodomethylene)adenosine 8b or its 5'-fluoromethylene analogue 11. Treatment of the 5'-aldehyde with hydroxylamine or dibromomethylene- or cyanomethylene-stabilized Wittig reagents and deprotections gave the oxime 4b, 5'-cyanomethylene 5b, and 5'-dibromomethylene 13b analogues. Dehydrobromination of 13b gave acetylenic compound 14b. From the tested 6-N-cyclopropyladenosine analogues modified at the 5' carbon, the 5'-iodomethylene 8b had the most potent activity against Trypanosoma brucei in vitro with an IC50 of 12 microg/mL. The IC50 value was 19 microg/mL for both the 5'-fluoromethylene 11 and the 5'-cyanomethylene 5b compounds. The (E)-5'-deoxy-5'-(iodomethylene)adenosine 2a, a known inhibitor of AdoHcy hydrolase not modified with a cyclopropyl ring at 6-amino group, also inhibited T. brucei with an IC50 of 9 microg/mL. In contrast to some other adenosine analogues modified at C5', the 6-N-cyclopropyladenosine analogues described here do not exhibit an inhibitory effect on AdoHcy hydrolase and displayed only marginal antiviral activity.


Subject(s)
Cyclopropanes/chemical synthesis , Deoxyadenosines/chemical synthesis , Trypanocidal Agents/chemical synthesis , Acetylene/analogs & derivatives , Acetylene/chemical synthesis , Acetylene/chemistry , Acetylene/pharmacology , Adenosylhomocysteinase/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Deoxyadenosines/chemistry , Deoxyadenosines/pharmacology , Humans , Oximes/chemical synthesis , Oximes/chemistry , Oximes/pharmacology , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects
20.
Arch Pathol Lab Med ; 130(3): 343-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16519562

ABSTRACT

CONTEXT: Clear cell sarcoma is a malignant soft tissue tumor with melanocytic differentiation. Molecular methods are sometimes necessary to identify the unique t(12; 22)(q13;q12) translocation and differentiate clear cell sarcoma from melanoma. OBJECTIVE: To determine whether CD117 immunoreactivity may be useful in separating melanoma from clear cell sarcoma. DESIGN: We identified 20 tumors listed in our surgical pathology files that were diagnosed as clear cell sarcoma or in which clear cell sarcoma was strongly considered. These were tested for the presence of the t(12;22) translocation by reverse transcriptase/polymerase chain reaction and sequencing from paraffin-embedded tissue. Tumors with a t(12;22) translocation were immunostained with an antibody to CD117 and compared with 16 similarly stained metastatic melanomas. RESULTS: Twelve tumors from 9 patients demonstrated t(12;22). No metastatic melanomas demonstrated t(12;22). None of the 12 clear cell sarcomas showed membrane or cytoplasmic staining for CD117. Conversely, 10 (63%) of 16 metastatic melanomas were, at least focally, positive for CD117; this difference was significant (P < .001). Interestingly, 3 tumors in which clear cell sarcoma was initially considered as a diagnosis, but which lacked t(12;22), were also positive for CD117. CONCLUSIONS: Reverse transcriptase/polymerase chain reaction, performed on paraffin-embedded tissue, is a useful, rapid tool for identifying the presence of t(12;22) in clear cell sarcoma. The CD117 immunoreactivity may prove useful in the differential diagnosis of deep soft tissue or visceral lesions with melanocytic differentiation; positive staining results exclude clear cell sarcoma, but are compatible with metastatic melanoma.


Subject(s)
Melanoma/diagnosis , Proto-Oncogene Proteins c-kit/analysis , Sarcoma, Clear Cell/pathology , Soft Tissue Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 22 , DNA, Neoplasm/analysis , Diagnosis, Differential , Female , Humans , Male , Melanoma/genetics , Melanoma/secondary , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Clear Cell/chemistry , Sarcoma, Clear Cell/genetics , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/genetics , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL